바로가기메뉴

본문 바로가기 주메뉴 바로가기

Regulation of CYP1A1 and Inflammatory Cytokine by NCOA7 Isoform 4 in Response to Dioxin Induced Airway Inflammation

Tuberculosis & Respiratory Diseases / Tuberculosis & Respiratory Diseases,
2015, v.78 no.2, pp.99-105







  • Downloaded
  • Viewed

Abstract

Background: Aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor, binds to a wide variety of synthetic and naturally occurring compounds. AhR is involved in the regulation of inflammatory response during acute and chronic respiratory diseases. We investigated whether nuclear receptor coactivator 7 (NCOA7) could regulate transcriptional levels of AhR target genes and inflammatory cytokines in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-treated human bronchial epithelial cells. This study was based on our previous study that NCOA7 was differentially expressed between normal and chronic obstructive pulmonary disease lung tissues. Methods: BEAS-2B and A549 cells grown under serum-free conditions were treated with or without TCDD (0.15 nM and 6.5 nM) for 24 hours after transfection of pCMV-NCOA7 isoform 4. Expression levels of cytochrome P4501A1 (CYP1A1), IL-6, and IL-8 were measured by quantitative real-time polymerase chain reaction. Results: The transcriptional activities of CYP1A1 and inflammatory cytokines were strongly induced by TCDD treatment in both BEAS-2B and A549 cell lines. The NCOA7 isoform 4 oppositely regulated the transcriptional activities of CYP1A1 and inflammatory cytokines between BEAS-2B and A549 cell lines. Conclusion: Our results suggest that NCOA7 could act as a regulator in the TCDD-AhR signaling pathway with dual roles in normal and abnormal physiological conditions.

keywords
Receptors, Aryl Hydrocarbon, Pulmonary Disease, Chronic Obstructive, Dioxins, NCOA7 Protein, Human

Reference

1.

1. Das SK. (2003), Harmful health effects of cigarette smoking. Mol Cell Biochem, 253,159-65.

2.

2. Kitamura M., Kasai A. (2007), Cigarette smoke as a trigger for the dioxin receptor-mediated signaling pathway. Cancer Lett, 252,184-94.

3.

3. Poland A., Knutson JC. (1982), 2,3,7,8-Tetrachlorodibenzo-p-dioxin and related halogenated aromatic hydrocarbons: examination of the mechanism of toxicity. Annu Rev Pharmacol Toxicol, 22,517-54.

4.

4. Lai H., Rogers DF. (2010), New pharmacotherapy for airway mucus hypersecretion in asthma and COPD: targeting intracellular signaling pathways. J Aerosol Med Pulm Drug Deliv, 23, 219-31.

5.

5. Barnes PJ. (2008), Immunology of asthma and chronic obstructive pulmonary disease. Nat Rev Immunol, 8,183-92.

6.

6. Mauad T., Dolhnikoff M. (2008), Pathologic similarities and differences between asthma and chronic obstructive pulmonary disease. Curr Opin Pulm Med, 14,31-8.

7.

7. Hylkema MN., Sterk PJ., de Boer WI., Postma DS. (2007), Tobacco use in relation to COPD and asthma. Eur Respir J, 29,438-45.

8.

8. Lai ZW., Hundeiker C., Gleichmann E., Esser C. (1997), Cytokine gene expression during ontogeny in murine thymus on activation of the aryl hydrocarbon receptor by 2,3,7,8-tetrachlorodibenzo- p-dioxin. Mol Pharmacol, 52,30-7.

9.

9. Vogel C., Abel J. (1995), Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin on growth factor expression in the human breast cancer cell line MCF-7. Arch Toxicol, 69,259-65.

10.

10. Vogel C., Donat S., Dohr O., Kremer J., Esser C., Roller M. (1997), Effect of subchronic 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure on immune system and target gene responses in mice: calculation of benchmark doses for CYP1A1 and CYP1A2 related enzyme activities. Arch Toxicol, 71,372-82.

11.

11. Warren TK., Mitchell KA., Lawrence BP. (2000), Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) suppresses the humoral and cell-mediated immune responses to influenza A virus without affecting cytolytic activity in the lung. Toxicol Sci, 56,114-23.

12.

12. Mimura J., Fujii-Kuriyama Y. (2003), Functional role of AhR in the expression of toxic effects by TCDD. Biochim Biophys Acta, 1619,263-8.

13.

13. Denison MS., Nagy SR. (2003), Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol, 43,309-34.

14.

14. Tojo M., Matsuzaki K., Minami T., Honda Y., Yasuda H., Chiba T. (2002), The aryl hydrocarbon receptor nuclear transporter is modulated by the SUMO-1 conjugation system. J Biol Chem, 277,46576-85.

15.

15. Beischlag TV., Wang S., Rose DW., Torchia J., Reisz-Porszasz S., Muhammad K. (2002), Recruitment of the NCoA/SRC-1/p160 family of transcriptional coactivators by the aryl hydrocarbon receptor/aryl hydrocarbon receptor nuclear translocator complex. Mol Cell Biol, 22,4319-33.

16.

16. Kasai A., Hiramatsu N., Hayakawa K., Yao J., Maeda S., Kitamura M. (2006), High levels of dioxin-like potential in cigarette smoke evidenced by in vitro and in vivo biosensing. Cancer Res, 66,7143-50.

17.

17. Gebremichael A., Tullis K., Denison MS., Cheek JM., Pinkerton KE. (1996), Ah-receptor-dependent modulation of gene expression by aged and diluted sidestream cigarette smoke. Toxicol Appl Pharmacol, 141,76-83.

18.

18. Shao W., Halachmi S., Brown M. (2002), ERAP140, a conserved tissuespecific nuclear receptor coactivator. Mol Cell Biol, 22, 3358-72.

19.

19. Paramanik V., Thakur MK. (2010), Interaction of estrogen receptor associated protein (ERAP) 140 with ER beta decreases but its expression increases in aging mouse cerebral cortex. Cell Mol Neurobiol, 30,961-6.

20.

20. Li H., Gomes PJ., Chen JD. (1997), RAC3, a steroid/nuclear receptorassociated coactivator that is related to SRC-1 and TIF2. Proc Natl Acad Sci U S A, 94,8479-84.

21.

21. Mangelsdorf DJ., Thummel C., Beato M., Herrlich P., Schutz G., Umesono K. (1995), The nuclear receptor superfamily: the second decade. Cell, 83,835-9.

22.

22. Naar AM., Beaurang PA., Zhou S., Abraham S., Solomon W., Tjian R. (1999), Composite co-activator ARC mediates chromatindirected transcriptional activation. Nature, 398,828-32.

23.

23. Norman AW., Litwack G. (1987), Estrogens and progestins. In: Litwack G, editor. Hormones. London: Academic Press. p. 550-60.

24.

24. Vogel CF., Matsumura F. (2009), A new cross-talk between the aryl hydrocarbon receptor and RelB, a member of the NF-kappaB family. Biochem Pharmacol, 77,734-45.

25.

25. Badal S., Delgoda R. (2014), Role of the modulation of CYP1A1 expression and activity in chemoprevention. J Appl Toxicol, 34,743-53.

26.

26. Lee AJ., Cai MX., Thomas PE., Conney AH., Zhu BT. (2003), Characterization of the oxidative metabolites of 17beta-estradiol and estrone formed by 15 selectively expressed human cytochrome p450 isoforms. Endocrinology, 144,3382-98.

27.

27. Ma X., Idle JR., Krausz KW., Gonzalez FJ. (2005), Metabolism of melatonin by human cytochromes p450. Drug Metab Dispos, 33,489-94.

28.

28. Androutsopoulos VP., Tsatsakis AM., Spandidos DA. (2009), Cytochrome P450 CYP1A1: wider roles in cancer progression and prevention. BMC Cancer, 9,187.

29.

29. Buterin T., Hess MT., Luneva N., Geacintov NE., Amin S., Kroth H. (2000), Unrepaired fjord region polycyclic aromatic hydrocarbon- DNA adducts in ras codon 61 mutational hot spots. Cancer Res, 60,1849-56.

30.

30. Rodriguez M., Potter DA. (2013), CYP1A1 regulates breast cancer proliferation and survival. Mol Cancer Res, 11,780-92.

Tuberculosis & Respiratory Diseases